59
Astrocytes in Parkinson’s disease

Astrocytes in Parkinson's disease

Embed Size (px)

DESCRIPTION

 

Citation preview

Page 1: Astrocytes in Parkinson's disease

Astrocytes in Parkinson’s disease

Master Thesis Neuroscience & CognitionIscha Bruinsma October – December 2009Supervisor: Simone van den Berge

Page 2: Astrocytes in Parkinson's disease

Abstract

Parkinson’s disease is a common neurodegenerative disorder with a high prevalence in people

over 65. Its main hallmark is progressive loss of dopaminergic neurons in the substantia nigra,

leading to characteristic motor symptoms such as tremor and bradykinesia. The pathology of

this disease is not yet fully understood. Recently it has been suggested that astrocytes might

play an important role in disease initiation and progression. In response to neuronal damage

astrocytes can enter a state called reactive astrogliosis, which is characterised by hypertrophy

and increased expression of glial fibrillary acidic protein. Though initially meant as a

protective reaction, this can have both beneficial and detrimental effects on surrounding

neurons. In this review the role that astrocytes might play in Parkinson’s disease is discussed.

Evidence of astrogliosis in animal models of Parkinson’s disease and post mortem studies of

patients is described, implicating the involvement of this process. Subsequently, several

factors are discussed that are important during the astrocytic response in Parkinson’s disease,

divided in neuroprotective and neurodegenerative effects. Based upon the astrocytic factors

that could contribute to disease pathology, some potential therapies for Parkinson’s disease

targeting astrocytes are suggested.

2

Page 3: Astrocytes in Parkinson's disease

Contents

Abstract.......................................................................................................................................2Contents......................................................................................................................................31. Introduction.............................................................................................................................4

1.1 Parkinson’s disease...........................................................................................................41.2 Animal models of PD........................................................................................................51.3 Astrocytes.........................................................................................................................81.4 Astrocytes in the substantia nigra.....................................................................................9

2. Astrogliosis...........................................................................................................................112.1 Astrogliosis in animal models of PD..............................................................................122.2 Astrogliosis in PD patients..............................................................................................132.3 Remote astrocyte activation............................................................................................142.4 Activation pathway.........................................................................................................14

3. Neurodegenerative & proinflammatory factors in astrocytes...............................................163.1 α-synuclein in astrocytes and Lewy bodies....................................................................163.2 ER stress and unfolded proteins......................................................................................173.3 The role of astrocytes in oxidative stress-induced neuronal degradation.......................173.4 MAO-B expression in astrocytes and production of reactive oxygen species................193.5 Immune responses and the role of astrocytes in sustaining inflammation......................203.6 Effect of hormones on astrocyte development and response to injury...........................21

4. Neuroprotective effects of astrocytes....................................................................................224.1 Protective effects of PAR-1 activation...........................................................................224.2 Role of Nurr1 in protecting dopaminergic neurons........................................................224.3 Anti-inflammatory and neuroprotective effects of hydrogen sulphide...........................234.4 The role of purines in regulation of the astrocytic response...........................................234.5 Astrocyte influence on microglia....................................................................................244.6 Protective effects of enriched environment....................................................................254.7 Astrocyte influence on neurogenesis..............................................................................254.8 Nrf2-mediated gene transcription inducing a large neuroprotective response...............26

5. Potential therapies for PD treatment targeting astrocytes.....................................................276. Discussion.............................................................................................................................29List of abbreviations.................................................................................................................32References.................................................................................................................................33

3

Page 4: Astrocytes in Parkinson's disease

Figure 1 In Parkinson’s disease neurons are lost in the substantia nigra, which is located in the midbrain. (howstuffworks.com)

1. Introduction

Though Parkinson’s disease was first described in 1817 its pathology is still not fully

understood. Several factors are known to contribute to disease initiation and/or progression

(Gasser 2009; Weidong Le et al. 2009) but it is not known why certain people develop the

disease while others do not. This review focuses on a particular aspect of the disease: the

involvement of astrocytes. The importance of astrocytes in normal brain function and their

possible involvement in neurodegenerative disorders has only recently been discovered. In

response to certain signals, astrocytes can start an active inflammatory response called

reactive astrogliosis (Eddleston & Mucke 1993), which is implicated to be important in the

pathology of Parkinson’s disease. Here, an overview is given of how astrocytes and reactive

astrogliosis might be involved in Parkinson’s disease and different models of this disease.

First, an introduction is given explaining some background information on Parkinson’s

disease, different animal models for this disease, and normal astrocyte function. Subsequently,

the concept of astrogliosis will be explained and some detrimental as well as beneficial factors

are presented that might play a role in reactive astrogliosis in Parkinson’s disease. Finally,

some potential therapies targeting astrocytes are suggested.

1.1 Parkinson’s disease

Parkinson’s disease (PD) is a common neurodegenerative disorder with a prevalence of 1.8 %

in people over 65 (Rijk et al.

2000). It is characterised by

disabling motor abnormalities

such as tremor, muscle rigidity,

bradykinesia, postural

instability and other

accompanying symptoms such

as fatigue and speech problems

(Purves et al. 2001) . Overall,

PD is primarily idiopathic with

only a subset of cases (<15% of

cases) with a family history of

4

Page 5: Astrocytes in Parkinson's disease

PD. In pedigrees with a pattern of inherited PD, genetic linkage studies have identified 13

PARK loci to date. Molecular genetics studies have identified genes associated with 7 of 13

PARK loci (Harvey et al. 2008). The primary neuropathological hallmark of PD is loss of

pigmented dopaminergic neurons in the substantia nigra pars compacta (SNpc), projecting to

the striatum. This results in a deficit in striatal dopamine levels, leading to most of the clinical

symptoms. In figure 1 the neuronal loss in the SN of a PD patient is shown compared to a

healthy individual. Until now, the most common treatment for this disease is administration of

levodopa (L-dopa), a dopamine precursor, which increases dopamine levels in the brain.

However, especially after chronic administration, this treatment has many side effects and

loses its effectiveness (Schapira et al. 2009). Moreover, L-dopa administration does not

interfere with disease progression but only alleviates the symptoms. Therefore, it is important

to require a deeper understanding of the causes and pathology of PD. Not only to try to

prevent the disease but also to develop new therapies that might stop or slowdown disease

progression.

PD is diagnosed pathologically by the loss of pigmented neurons in the SNpc and is

associated with widespread occurrence of Lewy bodies and dystrophic Lewy neurites

throughout the central and autonomic nervous system (Gelb et al. 1999). These are abnormal

intracellular aggregates of α-synuclein in respectively neurons and axons, that might

contribute to disease pathology. It has also been found that oxidative stress plays an important

role in disease pathology and might be the main cause of neuronal degradation. In post

mortem studies, evidence of increased oxidative stress in the SNpc of PD patients has been

observed (Hunot et al. 1996).

1.2 Animal models of PD

PD is a multifactorial disease with a complex etiology that results from genetic risk factors,

environmental exposures and most likely a combination of both. Rodent models of

Parkinsonism aim to reproduce key pathogenic features of the syndrome, including movement

disorder induced by the progressive loss of dopaminergic neurons in the substantia nigra,

accompanied by the formation of α-synuclein containing Lewy body inclusions. The two most

widely used models for PD are the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)

model and the 6-hydroxydopamine (6-OHDA) lesion model, these and other rodent models

are reviewed in Melrose et al. 2006.

5

Page 6: Astrocytes in Parkinson's disease

MPTP is a by-product of the chemical synthesis of a meperidine analogue with potent heroin-

like effects. It can induce a Parkinsonian syndrome in humans almost indistinguishable from

PD. MPTP is a potent and irreversible mitochondrial complex I inhibitor whose toxic

metabolite MPP+ is selectively transported by the dopamine transporter DAT (Watanabe et

al. 2005). The conversion of MPTP in astrocytes and the effect of MPP+ on neurons is shown

in figure 2. MPTP causes damage to the dopaminergic pathways identical to that seen in PD

with a greater loss of neurons in the SNpc than in the ventral tegmental area (Muthane et al.

1994) and greater loss of nerve terminals in the putamen than in the caudate nucleus

(Moratalla et al. 1992). Differences with PD are that MPTP damage has much faster effects

and no Lewy bodies are found after MPTP administration in humans, though there is

upregulation of α-synuclein expression in rodents and primates (Meredith et al. 2002).

Dopaminergic neurons in rats are relatively resistant to MPTP-induced toxicity and in mice

susceptibility of the nigrostriatal pathway to neurodegeneration is strain dependant (Muthane

et al. 1994).

6-OHDA administration causes nigrostriatal depletion when stereotaxically injected into the

SN, median forebrain bundle or striatum. It destroys catecholaminergic neurons through a

combination of reactive oxygen species and increased toxic quinones (reviewed by Bove et al.

2005). After injection dopaminergic neurons in the SNpc die within 24 hours, show apoptotic

Figure 1 Schematic representation of the conversion of MPTP in astrocytes and the effects of MPP+ on neurons (Vila & Predborski 2003).

6

Page 7: Astrocytes in Parkinson's disease

morphology and decreased α-synuclein mRNA (corresponding with neuronal degeneration),

creating a PD like situation. A difference with PD is that Lewy body formation has not been

observed in this model.

Models based on neurotoxins have enormous value in helping to understand the consequences

of nigrostriatal loss and to test symptomatic therapies and interventions. However, there is no

clear evidence that the mechanisms of action of these toxins are similar to neuropathological

processes occurring in PD. Furthermore, there is no evidence to date that effective

neuroprotection against these toxins translates into an effective neuroprotective therapy in

humans with PD. Another major limitation of the toxin-based models is that they do not

reproduce the pathology and cell loss observed in other brain regions and peripheral tissues of

patients, nor the broad range of non-motor symptoms seen in PD.

Next to models based on neurotoxicity, there are now also transgenic models available.

Genetic mutations identified in familial Parkinsonism have recently provided a new approach

to understand the molecular pathways affected. Transgenic models with knock-outs,

overexpression or mutations in single genes provide a powerful new set of molecular tools to

study etiology. Examples of genes that can be manipulated in PD models are recessively

inherited loss-of-function mutations in Parkin, DJ-1 and PTEN-induced putative kinase-1

(PINK1) (for review see Harvey et al. 2008). In the past decade these were found to cause

early-onset (< 50 years at onset), L-DOPA-responsive Parkinsonism. The slowly progressive

and predominant motor phenotype in these patients suggests a disorder largely restricted to

dopaminergic neuronal loss. The majority of patients with Parkin-linked disease demonstrate

neuronal loss restricted to the substantia nigra. In contrast, dominantly inherited, gain-of-

function mutations in α-synuclein and leucine-rich repeat kinase result in more typical, late-

onset, Lewy body Parkinsonism with multi-system involvement (Ross & Farrer 2005).

Presently, it is unknown whether genetic causes identified in rare, Mendelian forms of

Parkinsonism highlight pathways affected in idiopathic PD. Parkinson’s syndrome most likely

results from an intricate combination of gene and gene-environment interactions. The most

optimal model for studying PD pathology would be one more closely reflecting sporadic

forms of the disease. The only genetic abnormality to date for which good evidence exists in

favour of a link with sporadic PD, is overexpression of wild-type α-synuclein via duplication

or triplication of the gene (Lee & Trojanowski 2006). Furthermore, patients with sporadic

forms of the disease present with abnormal α-synuclein accumulation and aggregates in a

subset of central and peripheral neurons (Halliday et al. 2006). There are several transgenic

mice models overexpressing α-synuclein using various promoters and mutations. A problem

7

Page 8: Astrocytes in Parkinson's disease

with choosing a promoter, is that this often restricts expression to a specific area or specific

neurons, while models should mimic the broad but regionally selective α-synuclein pathology

observed in patients. It has also been found that some modifications, such as double mutation

or truncation, are necessary to obtain cell loss and decrease dopamine levels in mice

(reviewed by Chesselet 2008). The relevance of these mechanisms of α-synuclein toxicity to

sporadic PD remains unclear because patients do not have doubly mutated α-synuclein and

the phenotype induced by the truncated protein so far does not mimic that of PD. However,

there is a reasonable likelihood that these models share mechanisms that occur in sporadic PD

(Chesselet 2008). Mutations accelerate the formation of abnormal forms of the protein that

can also be adopted by wild-type α-synuclein, and truncated forms of the protein are found in

patient brains (Follmer et al. 2007). Another approach is to overexpress α-synuclein using

viral vectors which produces a rapid degeneration of nigrostriatal neurons (Kirik & Bjorklund

2003). This revealed the ability of wild-type α-synuclein to induce nigrostriatal pathology.

The well controlled regional and temporal overexpression, and the lack of expression during

embryonic and post-natal development, which may better mimic disease conditions and avoid

the upregulation of defence mechanisms, are distinct advantages. However, only a subset of

neurons is transduced in these models, which again brings up the problem of multiple affected

neuronal systems in PD pathology (Chesselet 2004).

Of course the shortcomings of available models should not discourage their use and delay

progress in the field of therapeutic strategies for PD. Each model has its own advantages and

disadvantages and they can be used to test various aspects of the disease.

1.3 Astrocytes

Astrocytes are the major cell population within the central nervous system (CNS), they make

up 55-60% of total brain cells. Astrocytes are complex highly differentiated cells that are

present throughout the entire CNS. They make numerous essential contributions to normal

functioning in the healthy CNS. Examples of their many functions are regulation of blood

flow, provision of energy metabolites to neurons, participation in synaptic function and

plasticity and maintenance of the extracellular balance of ions, fluids and transmitters. Other

active functions might be synchronisation of neuronal firing patterns through neurotransmitter

release (reviewed by Volterra 2005).

8

Page 9: Astrocytes in Parkinson's disease

Astrocytes express glial fiblillary acidic protein (GFAP) which is an intermediate filament

involved in controlling the shape and movement of astrocytes and is important for astrocyte-

neuronal interaction. GFAP-mediated astrocytic processes play a vital role in modulation

synaptic efficacy in the CNS. GFAP is also essential for normal white matter architecture and

blood-brain barrier integrity (reviewed by Eng et al. 2000).

Not too long ago, it was believed that astrocytes were inactive elements just providing a

scaffold function in the CNS. Now, it is known that astrocytes express almost the same set of

ion channels and receptors as neurons and they can respond to activation and have active

modulatory roles in intercellular communication (Seifert et al. 2006; Volterra 2005). It was

also found that not all astrocytes are the same with respect to antigen profiles and functional

properties. However, not much is known yet about expression and function of these different

astrocytes.

1.4 Astrocytes in the substantia nigra

It has been known for a long time that not all astrocytes in the brain are the same. Initially, a

difference was found between astrocytes in grey and white matter but later it was discovered

that there is also regional and even intraregional heterogeneity (Bachoo et al 2004). In the

brain, astrocytes have an ordered arrangement with minimal overlap, forming discrete

territories in parallel with neuronal and vascular territories (Bushong et al. 2002). Astrocytes

in specific brain areas might differ for example in the neurotransmitter receptors they express,

their immune response, opiod receptor expression or gap junction coupling (Yeh et al. 2009).

Astrocytes can also be divided in one half exhibiting high GFAP expression, low input

resistance, a typical irregular cell body with branched processes and low membrane potential

and another half with low GFAP expression, larger input resistance and lower glutamate

uptake that are not couple through gap junctions (Volterra et al. 2005).

Though it is widely known that astrocytes form a heterogeneous population, not much is

known about specific properties of astrocytes in certain brain areas such as the substantia

nigra, which is important in PD. The reason for the specific loss of dopaminergic neurons in

the SN in PD might be based on special properties of the astrocytes in this area. These could

be more vulnerable to mutations or susceptible to environmental effects, and could have a

different response to injury than astrocytes in other brain areas. It is known, for example, that

there are lower numbers of astrocytes in the SN than in other brain areas (Mena & Yebenes

9

Page 10: Astrocytes in Parkinson's disease

2008). If the dopamine neurons, which spontaneously generate abundant free radicals during

the metabolism of dopamine, are less protected by a smaller proportion of guardian cells with

high free radical scavenging properties, such as the astrocytes, then this could be an

explanation for the increased susceptibility of the nigral dopamine neurons. Astrocytes

support the differentiation, survival, pharmacological properties, and resistance to injury of

dopamine neurons (Mena & Yebenes 2008).

Astrocytes in the striatum have also been found to express relatively high levels of

intercellular adhesion molecule-1 (ICAM-1), an inflammatory mediator (Morga et

al. 1998), which might make this region more susceptible to inflammation.

The SN appears to be particularly vulnerable to inflammatory processes. For example,

lipopolysaccharide (LPS), an inducer of immune response, injected into the SN leads to loss

of tyrosine hydroxylase(TH)-cells, while there is no cell loss when it is injected into the

hippocampus or cortex (Liu et al. 2006). This vulnerability could be due to the high level of

oxidative action in dopaminergic neurons or to the possible higher abundance of microglial

cells.

Another factor making the SN more vulnerable is the high levels of MAO-B in this area in

neurons and astrocytes (Damier et al. 1996). MAO-B metabolises dopamine, which is present

in particularly high levels in the SN, because it is produced here, and is released by damaged

neurons. During conversion of its substrate, MAO-B produces ROS, which increases

oxidative stress and can be damaging to neighbouring neurons.

10

Page 11: Astrocytes in Parkinson's disease

Figure 3 Photomicrographs of immunohistochemical staining of glial fibrillary protein (GFAP) in astrocytes in wild type mice. In healthy tissue and of different gradations of reactive astrogliosis and glial scar formation after tissue insults of different types and different severity. (Sofroniew 2009)

2. Astrogliosis

Reactive astrogliosis, whereby astrocytes undergo varying molecular and morphological

changes, is a poorly understood hallmark of all central nervous system pathologies. Reactive

astrogliosis can be induced by all forms and severities of CNS injury or disease including

subtle perturbations. It is not an all-or-nothing response but a finely gradated continuum of

progressive changes in gene expression and cellular changes that are subtly regulated by

complex intercellular and intracellular signalling. The changes undergone by reactive

astrocytes vary with the nature and severity of the injury and are regulated in a context

specific manner (Sofroniew 2009). Cellular changes include hypertrophy, and in severe cases,

proliferation and scar formation, as can be seen in figure 3 and 4. A very important aspect of

astrogliosis is the upregulation of GFAP, which is often used as a marker for reactive

astrogliosis (Eddleston & Mucke 1993). In figure 3 an immunohistochemical staining for

GFAP is shown in healthy tissue and different severities of astrogliosis, clearly showing

morphological changes during reactive astrogliosis. The changes undergone during reactive

gliosis have the potential to alter astrocyte activity both through loss and gain of functions

(Sofroniew 2009). These changes can have both beneficial and detrimental effects on

surrounding neural and non-neural cells. Several aspects of reactive astrogliosis might play a

role in PD pathology. Though astrogliosis might be beneficial in many ways, for example in

maintaining extracellular glutamate levels and homeostasis in the striatum after dopaminergic

neuronal loss, normal astrocytic functions might be compromised during astrogliosis. It has

been found for example that the number of glutamate transporters per astrocyte is reduced in a

model of chronic PD (Dervan et al. 2004). Changes in astrocyte ability to regulate glutamate,

11

Page 12: Astrocytes in Parkinson's disease

and its associated synaptic functions, could be important for the progressive nature of the

pathophysiology associated with Parkinson’s disease.

Figure 2 schematic overview of changes undergone by astrocytes during reactive astrogliosis. (Buffo et al. 2009)

2.1 Astrogliosis in animal models of PD

In the MPTP model, the start of an astrocytic reaction is observed two days after MPTP

application (Kohutnica et al. 1998). This is one day later than the start of the microglial

reaction, suggesting that the astrocytic reaction depends on factors released by microglia, such

as interleukin-1, which is a known stimulator of astrogliosis (Giulian & Lachman 1985). In

the striatum the response is maximal after 5 days and in the SN 14 days after MPTP

administration (Kohutnica et al. 1998). Blocking conversion of MPTP into MPP+ with

pergyline, a MAO-B inhibitor, inhibits the astrocytic response to MPTP. Although the

conversion of MPTP to MPP+ occurs mainly in astrocytes, MPTP alone is not a factor

inducing the reaction of these cells. In MPTP induced reactive astrocytes, interleukin-6 (IL-6)

expression has also been detected (Kohutnica et al. 1998). It has been shown that IL-6 induces

the synthesis of neurotrophic factors, such as nerve growth factor, by astrocytes (Frei et al.

1989), and inhibits the production of neurotoxic agents like tumor necrosis factor α (TNFα)

(Aderka et al. 1989). Overexpression of this cytokine leads to an increase in the number of

GFAP positive astrocytes (Fattori et al. 1995). Furthermore, in the MPTP model, astrocytic

12

Page 13: Astrocytes in Parkinson's disease

activation parallels the time course of dopaminergic cell death in the SNpc as well as striatum

and GFAP expression remains upregulated, even after most of dopaminergic neurons have

died due to MPTP intoxication. These findings implicate that astrocytic reaction occurs after

neuronal cell death and might play a role in the propagation of the neurodegenerative process

but not in its initiation (Liberatore et al. 1999).

In the 6-OHDA model, upregulation of GFAP is seen 24 hours after lesioning, and peaks at 4

days when GFAP levels are almost 4 times as high. This astrocytic response is transient and

returns to control levels after 28 days. When 6-OHDA is injected unilaterally a small

astrocytic response can be seen in the SN at the controlateral site as well. Furthermore, 6-

OHDA injection can induce GFAP changes over long distances in the striatum and even in the

cortex (Henning et al. 2008; Sheng et al. 1993). These results suggest that astroglial reaction

is triggered directly or indirectly by factors released from damaged neurons. However, it

might be that astroglial reaction is triggered only by the injection lesion and not specifically

by 6-OHDA damage (Depino et al. 2003).

2.2 Astrogliosis in PD patients

Though reactive astrogliosis can readily be found in animal models of PD, autopsy studies of

patients clinically and pathologically diagnosed with PD suggest that there might not be any

significant astrogliosis in the substantia nigra of PD patients. Some immunohistochemistry

experiments on the SN and putamen of PD patients did not find any alterations in the amount

of GFAP-immunoreactive astrocytes or in their morphology, compared to control brains.

Typical morphological characteristics of astrogliosis, such as hyperthrophy, shortening of

cytoplasmic processes and nuclear enlargement, were not found. Moreover, no difference in

the expression of metallothioneins was found, which are normally increased as protection to

increased oxidative stress (Mirza et al. 2000). However, others did find increased amounts of

reactive astrogliosis in PD patients, using GFAP-immunoreactivity (Miklossy et al. 2006).

This reactive astrogliosis was accompanied by high inter-cellular adhesion molecule 1(ICAM-

1) expression in the astrocytes and high expression of the counterreceptor Lymphocyte

function-associated antigen 1 (LFA-1) in microglia,which might be important for sustaining

inflammation (as explained in section 3.5).

At post mortem examinations it has also been found that there are increased amounts of NO

radicals in brains of PD patients (Hunot et al. 1996). In addition to evidence for increased NO

production in PD, impairment of mitochondrial function is also evident (Heales et al. 2004). It

13

Page 14: Astrocytes in Parkinson's disease

has also been shown that gluthatione, an important antioxidant enzyme, is downregulated in

the SN of PD patients (Pearce et al. 1997; Sian et al. 1994)

2.3 Remote astrocyte activation

In PD and animal models of PD, astrocytes are not only active at the site of

neurodegeneration, the SN, but also in other areas such as the subthalamic nucleus (STN) and

globus pallidus (GP)(Henning en al. 2008). It is hypothesised that communication links exist

between astrocytes, or between neurons and astrocytes, along neuronal pathways that transmit

activating signals in response to neuronal damage, but only if the neuronal pathways are at

least partially intact. Analysis of astrocyte activation in two 6-OHDA rat models of PD:

partial and complete SNc lesions, by injections of 6-OHDA in the striatum and medial

forebrain bundle, respectively, has led to the finding that astrocyte activation after partial

lesioning can spread to the GP and STN while complete lesioning results only in astrocyte

activation at the lesion site (Henning et al. 2008). Astrocytes can presumably propagate

information about neuronal damage or reduced activity through gap-junction linked astrocytic

networks. Complete degradation of a neuronal pathway might lead to breakdown of glial

communication, for example through loss of gap-junctions, which are no longer necessary for

uptake of excess ions or neurotransmitters.

2.4 Activation pathway

Though reactive gliosis is a very common phenomenon not much is known about the

molecular pathways leading to this reactive state in various neurodegenerative diseases.

Analysis of gene expression changes and protein phosphorylation in the MPTP model has

identified the JAK-STAT pathway to be involved in astrocyte activation in PD.

Administration of MPTP caused rapid phosphorylation by JAK2 and nuclear translocation of

STAT3 in striatal astrocytes, prior to the induction of GFAP mRNA and protein (Sriram et al.

2004). Phosphorylated STAT3 can enhance transcription of GFAP possibly via a STAT3

binding site in the GFAP promoter. This indicates that the JAK2/STAT3 pathway is involved

in induction of astrogliosis. The JAK2/STAT3 can be activated by several gp130-related

cytokines, such as IL-6, leukaemia inhibitory factor (LIF) and oncostatin-M (OSM).

Expression of these ligands can be induced by MPTP-mediated neuronal damage, which

14

Page 15: Astrocytes in Parkinson's disease

could be the trigger for astrogliosis (Sriram et al. 2004). A schematic representation of this

pathway is given in figure 5.

15

Page 16: Astrocytes in Parkinson's disease

Figure 5 Schematic diagram showing the involvement of gp130-mediated phosphorylation of JAK2/STAT3 pathway inducing GFAP expression. Upon putative ligand (e.g. IL-6, LIF, OSM) binding, JAK2 and STAT3 are recruited to the gp130-signal transducer, JAK2 phosphorylates the Tyr-705 residue on STAT3. The phosphorylated STAT3 dimerize, translocate to the nucleus and mediates transcriptional activation of astrocytic genes such as GFAP. (Sriram et al. 2004)

3. Neurodegenerative & proinflammatory factors in astrocytes

Reactive astrogliosis in response to injury is a mechanism to clean up damage and control its

spread. Part of this reaction is the stimulation of inflammatory processes which can help

attract other damage controlling cells. However, too much inflammation increases, instead of

restricts damage. In some situations, the immune system can start to attack healthy cells

instead of just cleaning up damaged ones. Astrocytes can stimulate such neurodegenerative

processes and sustain the inflammatory response, sometimes even after the initial trigger is

gone.

16

Page 17: Astrocytes in Parkinson's disease

3.1 α-synuclein in astrocytes and Lewy bodies

α-synuclein (α-syn) is a 14 kDa acidic protein concentrated in presynaptic neuronal terminals

(Norris et al. 2004). Normal physiological functions are hypothesised to include synaptic

vesicle turnover, synaptic plasticity, ubiquitin-proteasome processing and molecular

chaperoning. Its mutations are a cause of autosomal dominant PD. Currently there are three

known point mutations in the α-syn gene: A30P, E46K and A53T; duplications and

triplications also play a role in PD development (Lee & Trojanowski 2006). WT and mutated

forms of α-syn can up-regulate ICAM-1 expression and IL-6 secretion in human astrocytes.

Mutated forms are more potent for this than WT forms, which might explain why they induce

autosomal dominant PD (Klegeris et al. 2006).

Aggregated α-syn is a major component of Lewy bodies, which are frequently found in PD

(Gelb et al. 1999), also in astrocytes. Astrocytic inclusions in other pathologies, such as

progressive supranuclear palsy and corticobasal degeneneration, usually contain tau and not

α-syn. Ultrastructurally astrocytic inclusions in PD are composed of a meshwork of randomly

arranged, loosely packed α-syn filaments, with diameters of 20-40 nm (Wakabayashi et al.

2000). The distribution of glial cells with inclusions in PD is similar to that of

catecholaminergic neurons in the midbrain. The amount of α-syn positive glial inclusions

correlates with nigrostriatal neuronal loss (Wakabayashi et al. 2000). Overexpression of α-syn

in astrocytes results in cell death (Stefanova et al. 2001) but α-syn aggregation in glial cells

takes a long time and degeneration of glial cells is much slower than that of neuronal cells

(Wakabayashi et al. 2000).

Unaggregated α-syn is an effective stimulator of astrocytes and inflammatory processes.

Leakage or excretion of this protein from normal or damaged neurons into the extracellular

space could potentially stimulate astrocytes into an inflammatory state. Monomeric and

aggregated α-syn may be secreted via a special endoplasmic/Golgi-independent exocytosis

pathway (Lee et al. 2005). Such secretion is enhanced by proteasomal and mitochondrial

dysfunction which have been found in PD.

α-syn could influence astrocytes through all three major MAPK pathways: ERK1/2, JNK and

p38, all of which have been associated with the actions of α-syn. Inhibitors of these MAPK

pathways lower IL-6 secretion and ICAM-1 expression in human astrocytes and α-syn

significantly increases phosphorylation of ERK1, ERK2, p47 JNK and p38 MAPK (Klegeris

et al. 2006).

17

Page 18: Astrocytes in Parkinson's disease

3.2 ER stress and unfolded proteins

It has been recently found that endoplasmic reticulum (ER) stress and aberrant protein

degradation might also play an important role in the pathogenesis of neurodegenerative

disorders. Homocysteine-induced endoplasmic reticulum protein is a stress-response protein

located in the ER membrane of neurons and astrocytes that can help defend against ER stress.

In PD this protein is found in neurons and astrocytes in the SN and it is accumulated in Lewy

bodies, suggesting a role in their formation. Unfolded proteins in astrocytes could induce the

inflammatory response (Slodzinski et al. 2009).

3.3 The role of astrocytes in oxidative stress-induced neuronal degradation

When astrocytes are stimulated by inflammatory mediators such as cytokines and

lipopolysaccharides, induction of the Ca2+ independent isoform of inducible nitric oxide

synthetase (iNOS), leads to generation of nitric oxide (NO) (Bolanos et al. 1994). In situations

of neuroinflammation, such as PD, astrocytes might contribute to neuronal cell death by

increasing NO production. NO also induces damage to the electron transport chain of brain

mitochondria leading to mitochondrial impairment, resulting in more oxidative stress

(Bolanaos et al. 1994). NO reacts with O2- to form peroxynitrite and its reactive intermediates

which can react indiscriminately with proteins, DNA, and other cell constituents.

On the other hand, upon exposure to NO, astrocytes increase their cellular glutathione (GSH)

availability, which can react with reactive oxygen species such as NO, reducing NO levels.

This can protect against oxidative stress induced neuronal degradation, which might be a

major cause of neuronal cell loss in PD. On top of this, increased astrocytic GSH levels lead

to increased GSH release, increasing neuronal protection. NO exposure leads to increased

GSH efflux from astrocytes, even after removal of NO. This is not caused by increased

permeability of the plasma membrane but is a regulated response (Gegg et al. 2003). Neurons

co-cultured with astrocytes approximately double their GSH concentration, protecting them

against NO damage (Gegg et al. 2003). Thus, astrocytes can be both protective against

oxidative stress by increasing their GSH production and increase oxidative stress via

production of NO, damaging neurons.

18

Page 19: Astrocytes in Parkinson's disease

From post mortem brain studies it is known that NO overproduction is found in PD patients.

Glial cells marked for high NOS activity and NO radicals have been

identified in the SN post mortem examination (Hunot et al. 1996).

Furthermore, nitrotyrosine (an index of reactive nitrogen species formation) residues and

elevated nitrosylated proteins have been reported, and determination of nitrite in cerebrospinal

fluid also implies increased formation of NO within the CNS (Heales et al. 2004). This

supports the damaging effects of astrocytes producing NO.

Proof of the importance of astrocytic gluthatione for neuronal survival comes from a study

using astrocyte-coated dialysis membranes placed directly on top of neuronal cultures to

provide a removable astrocyte layer between the neurons and the culture medium (Chen et al.

2001). Using this technique, it was established that astrocytes can protect neurons against NO

induced damage. Furthermore, it was found that gluthatione-depleted astrocytes cannot

protect neurons against NO damage and the gluthatione content of astrocytes directly reflects

their protective potential. GSH has a special function in protection against NO, but does not

influence normal neuronal survival in the absence of NO. Because astrocyte processes form a

nearly continuous membrane around neuronal cell bodies and processes, they can reduce the

amount of NO reaching neurons by trapping it. GSH can also function as an exchange of

reducing equivalents between astrocytes and neurons.

Unfortunately, it is known that the protection afforded to neurons by astrocytes is finite,

possibly due to a decline in GSH trafficking with chronic iNOS induction (Heales et al. 2004).

Depletion of GSH may facilitate production of reactive oxygen species by astrocytes. GSH is

important in limiting and repairing the deleterious actions of NO, but GSH levels can be

depleted by too high concentrations of NO (Chen et al. 2001). In PD a 40% loss of GSH has

been reported in the SN (Sian et al. 1994). This is at odds with elevated GSH as a protection

mechanism, but could be explained by the mechanism being transient. This decrease in GSH

concentration precedes other hallmarks of PD and may be important in the early pathogenesis

of PD.

The reason that mainly neurons are affected in PD could be that neurons seem to be more

susceptible to NO exposure. GSH concentration is normally higher in astrocytes than in

neurons and in neurons NO exposure leads to a decrease instead of an increase in GSH. There

is also a greater inhibition of the mitochondrial respiratory chain in neurons than in astrocytes

exposed to NO (Gegg et al. 2003). GSH is synthesised by glutamate-cysteine ligase (GCL),

which is the rate limiting enzyme. The increased GSH concentration in astrocytes might be

explained by increased GCL activity after NO exposure, while in neurons there is no change

19

Page 20: Astrocytes in Parkinson's disease

in this activity. The high sensitivity of neurons to NO might be caused by low CGL activity

and the inability to increase this activity upon NO exposure, which leads to low GSH

concentrations and greater susceptibility to oxidative stress.

Increased iNOS expression by astrocytes, which produces excessive amounts of NO, could

also play a role in inducing increased GFAP expression, a hallmark of reactive astrogliosis.

Inflammatory mediators and inducers of NO production induce the expression of GFAP in

astrocytes via NO. This induction is inhibited by scavenging NO or inhibiting iNOS, while

NO alone is sufficient to stimulate the expression of GFAP in astrocytes, independent of

microglia. It has been found that the expression of GFAP in astrocytes can be increased via a

NO-GC-cGMP-PKG pathway (Brahmachari et al. 2006), which might be important under

neurodegenerative conditions, for example in PD, and could be a pharmacological target for

new therapies.

3.4 MAO-B expression in astrocytes and production of reactive oxygen species

Monoamineoxidase-B (MAO-B) is found in the brain primarily in non-neuronal cells such as

astrocytes and radial glia. The SN contains especially high levels of MAO-B positive

astrocytes, which might play an important role in PD pathology (Damier et al. 1996). MAO-B

levels are known to increase with age and in association with neurodegenerative diseases

(Kumar & Andersen 2004).

During oxidation of its substrate, this enzyme reduces oxygen to H2O2, which is a reactive

oxygen species (Cohen et al. 1997). It has been postulated that age-related increases in MAO-

B activity may contribute to cellular degeneration in the brain due to corresponding increases

in the production of this reactive oxygen species. Astrocytes themselves are protected against

the H2O2 they produce because they contain high levels of GSH and gluthatione peroxidase,

which can detoxify H2O2 within cells. Surrounding neurons however, are very vulnerable

because they contain lower levels of these protective agents, as explained in section 3.3. High

amounts of H2O2 produced in astrocytes, expressing high amounts of MAO-B, can diffuse to

neurons and contribute to mitochondrial damage and neuronal cell death. MAO-B activity

levels have been found to be doubled in the SN in Parkinson’s disease, and to correlate with

the percentage of dopaminergic SN cell loss (Damier et al. 1996). In vitro studies have shown

that induced increases in astrocytic MAO-B levels result in specific inhibition of

20

Page 21: Astrocytes in Parkinson's disease

mitochondrial complex 1 activity in cultured dopaminergic cells (Kumar et al. 2003).

Selective reductions in complex 1 activity have also been associated with PD.

It has even been shown that elevations in astrocytic MAO-B result in a relatively selective

loss of dopaminergic neurons in the SN (Mallajosyula et al. 2008). This cell loss was

accompanied by increased mitochondrial oxidative stress and selective decreases in complex

1 activity along with local microglia activation. These pathological findings correlated with a

significant decrease in locomotor activity. Thus, increased MAO-B activity is able to induce

several pathological hallmarks of PD (Mallojosyula et al. 2008). This suggests that MAO-B

may be a common initiator for these events and provides a novel model for exploring the

mechanisms by which these events can occur in the context of the human condition.

3.5 Immune responses and the role of astrocytes in sustaining inflammation

Several lines of evidence suggest that inflammatory mediators such as TNFα, NO and

interleukin-1, derived from microglia and astrocytes modulate the progression of PD

(Teisman & Shulz 2004). Environmental factors, such as infection, may interact with common

but less penetrant susceptibility genes to influence the onset of most commonly observed

sporadic PD cases.

Neuroinflammation in the SN, once initiated, may be self-sustaining, and the SN seems to be

particularly vulnerable to inflammation (Klegeris & McGeer 2007). An important

contributing protein in inflammation of the SN in PD cases could be ICAM-1. ICAM-1 with

its counter receptor LFA-1, is known to play a key role in inflammatory processes. In patients

with neuropathologically confirmed PD, high numbers of ICAM-1 positive reactive astrocytes

were found (Miklossy et al. 2006). In the SN, these ICAM-1 positive astrocytes were

concentrated around residual neurons in areas of heavy neuronal loss. LFA-1 positive

microglia gathered in such areas of high ICAM-1 expression, and LFA-1 leukocytes

infiltrated the tissue (Miklossy et al. 2006). ICAM-1 and LFA-1 are known to play a key role

in setting the level of inflammation in many other inflammatory diseases, such as rheumatoid

arthritis, coronary heart disease, type 1 diabetes and Alzheimers disease (see Miklossy et al.

2006). It is possible that overexpression of ICAM-1 with its ligand LFA-1 in PD patients is

responsible for sustaining inflammation in the SN and that this process is responsible for

autodestruction of SN dopaminergic neurons (Klegeris et al. 2006).

21

Page 22: Astrocytes in Parkinson's disease

Figure 6 Schematic overview of the effects of estrogen and glucocorticoids on neurons and astrocytes (Marchetti et al. 2005)

3.6 Effect of hormones on astrocyte development and response to injury

Exposure to hormones is another factor that could influence disease predisposition and

severity, possibly through an effect on astrocytes. Differences in hormone exposure might

also explain differences in PD prevalence between males and females. Alterations in

developmental programming of neuroendocrine and immune system function may critically

modulate vulnerability to Parkinson’s disease. It was found that hormonal programming has

an important effect on glial response to inflammation and oxidative stress. Studies using

glucocorticoid-deficient and estrogen-deprived mice showed that endogenous glucocorticoids

and the female hormone estrogen inhibit the aberrant neuroinflammatory cascade, protect

astrocytes and microglia from programmed cell death, and stimulate recovery of dopamine

neuron functionality, thereby triggering the repair process (Marchetti et al. 2005). In figure 6 a

schematic overview can be seen of the effects of estrogen and glucorticoids on activated

astrocytes.

22

Page 23: Astrocytes in Parkinson's disease

4. Neuroprotective effects of astrocytes

Of course, reactive astrogliosis does not happen without a reason. Initially, it is meant to have

a protective instead of a neurodegenerative effect. Just like astrocytes support neurons during

normal brain function, they continue to offer this support in situations of neuronal damage.

Moreover, they can stimulate repair via neurotrophic factors and control the inflammatory

response, preventing it from becoming excessive.

4.1 Protective effects of PAR-1 activation

A factor that is increased in reactive astrocytes in the SNpc of PD patients is protease

activated receptor 1 (PAR-1), a thrombin receptor. Thrombin-mediated activation of human

astrocytes results in morphologic changes and increased proliferation characteristic of reactive

astrogliosis. Moreover, this results in an increase in glial cell line-derived growth factor and

gluthatione peroxidase but not in inflammatory cytokines. Gluthatione peroxidase release

from these activated astrocytes has a protective effect on surrounding neurons (Ishida et al.

2006). Increased expression of PAR-1 seems to be a restorative move to protect against

neurotoxicity.

4.2 Role of Nurr1 in protecting dopaminergic neurons

Nurr 1 belongs to the nuclear receptor (NR)4 family of orphan nuclear receptors and is known

to function as a constitutively active transcription factor by binding to target genes as a

monomer, homodimer or heterodimer with other receptors (Wang et al. 2003). It has an

essential role in development and/or maintenance of dopaminergic neurons. Human mutations

resulting in reduced expression of Nurr 1 are associated with late-onset familial PD,

indicating that normally Nurr 1 may play a protective role (Le et al. 2003).

Indeed it was found that Nurr 1 plays a previously unexpected role in protecting dopaminergic

neurons from inflammation-induced neurotoxicity (Saijo et al. 2009). It functions as an

inhibitor of inflammatory gene expression in microglia and astrocytes. Astrocytes can act as

amplifiers of microglia-derived mediators in the production of neurotoxic factors. Nurr 1

protects the CNS from amplification of inflammatory signalling via microglia-astrocyte

communication. Anti-inflammatory activity is mediated by a Nurr1/CoREST transpression

pathway that operates in a feedback manner to restore transcription of NF-κB target genes to a

23

Page 24: Astrocytes in Parkinson's disease

basal state. NF-κB target genes include many pro-inflammatory proteins. Nurr1 is recruited to

NF-κB on inflammatory gene promoters via several mediators. It is suggested that the

CoREST transpression pathway might be widely used by members of the NR4A family.

Reduction of most of the well-established components of the CoREST complex severely

compromises the anti-inflammatory activity of Nurr1. Defects in the expression or activity of

these proteins could predispose individuals to PD (Saijo et al. 2009).

4.3 Anti-inflammatory and neuroprotective effects of hydrogen sulphide

Hydrogen sulphide (H2S) is a physiological product generated by all the tissues in the body

and a high production in the brain. It has anti-inflammatory and neuroprotective effects and is

also a powerful antioxidant, both directly and indirectly, by keeping other antioxidants such as

GSH in a reduced state. It can also react with nitrite ions, neutralising iNOS activity. Both

iNOS activity and oxidation products are known to be important in PD pathogenesis,

suggesting possible involvement of H2S.

In the brain, H2S is synthesised from L-cyteine, via cystathionine-b-synthase (CBS).

Astrocytes are the most powerful producers of H2S in the brain, and most strongly express

CBS, as was found in post mortem brain tissue (Lee et al. 2009). Inflammatory stimulation

causes a reduction in this CBS expression, and thus in H2S anti-inflammatory activity.

Endogenous H2S production has a protective effect against release of inflammatory mediators

by stimulated astrocytes, this can be stimulated by exposure to SH ions (from H2S in

solution). NaSH treatment and endogenous H2S have neuroprotective effects through

reduction of toxic materials secreted by glial cells subjected to inflammatory stimuli, and

reduce production of inflammatory mediatiors including NO (Lee et al. 2009). Externally

supplied SH can also reduce activation of NF-κB which is normally induced by inflammatory

stimulation of astrocytes, and suggested to be involved in PD (Saijo et al. 2009).

4.4 The role of purines in regulation of the astrocytic response

Purines also play a significant role in the pathophysiology of numerous acute and chronic

disorders of the central nervous system and might play a role in PD. This hypothesis is

supported by the protective effect of adenosine 2A receptor antagonist in PD models and

epidemiological studies (Jenner et al. 2009). Astrocytes are the main source of cerebral

24

Page 25: Astrocytes in Parkinson's disease

purines (Cicarelli et al. 1999). They release either adenine-based purines, e.g. adenosine and

adenosine triphosphate, or guanine-based purines, e.g. guanosine and guanosine triphosphate,

in physiological conditions and release even more of these purines in pathological conditions.

Astrocytes express several receptor subtypes of types for adenine- and guanine-based purines

(Cicarelli et al. 2001). Specific enzymes metabolise both adenine- and guanine-based purines

after release from astrocytes. This regulates the effects of nucleotides and nucleosides by

reducing their interaction with specific membrane binding sites. Adenine-based nucleotides

stimulate astrocyte proliferation via an increase in intracellular [Ca2+] and specific effects on

proteins. Adenosine also may stimulate astrocyte proliferation, but mostly inhibits astrocyte

proliferation, thus controlling excessive reactive astrogliosis. The activation of certain

adenosine receptors also stimulates astrocytes to produce trophic factors, which contribute to

protect neurons against injuries. Guanosine stimulates the output of adenine-based purines

from astrocytes and in addition it directly triggers these cells to proliferate and to produce

large amount of neuroprotective factors. These data indicate that adenine- and guanine-based

purines released in large amounts from injured or dying cells of CNS may act as signals to

initiate brain repair mechanisms widely involving astrocytes (Ciccarelli et al. 2001).

4.5 Astrocyte influence on microglia

Although inflammation is an indispensable defense mechanism against pathogens, it often

damages surrounding tissues. Therefore, the extent of inflammation should be tightly

controlled to maximize the antipathogenic effect, while minimizing tissue damage. Astrocytes

might play an important role in this because they can modulate the activity of microglia,

which are responsible for the biggest part of the inflammatory response in PD.

Reactive oxygen species (ROS) are one of the major signalling molecules capable of

modulating microglia activation (Min et al. 2003). Therefore, microglial activation could be

regulated by modulating intracellular ROS level. A candidate molecule to regulate

intracellular ROS is the antioxidant enzyme heme oxygenase-1 (HO-1), which also has anti-

inflammatory effects. Min et al. (2006) demonstrated that astrocyte culture conditioned

medium (ACM) enhance HO-1 expression and activity in microglia. Furthermore, treatment

with ACM suppressed interferon-γ-induced ROS production, leading to reduced iNOS

expression and NO release.

25

Page 26: Astrocytes in Parkinson's disease

4.6 Protective effects of enriched environment

It has been found that enriched environment (EE) has protective effects

against neurodegeneration (Anastatasia et al. 2009). EE is defined as a

sustained and progressive increase in cognitive and sensorimotor stimuli, with aggregated

voluntary physical activity and complex social interactions. EE significantly reduces 6-

OHDA degeneration of dopaminergic neurons in the SNpc of adult rats, preserves

nigrostriatal projections, and most importantly, improves the dopaminergic function. EE

resulted in a marked increase in GFAP expression in the SN after 6-OHDA lesioning

(Anastasia et al. 2009). This suggests that an early post-lesion astrocytic reaction may

participate in the neuroprotective mechanism. It appears that animals exposed to an EE have

an increased ability to respond to the toxic injury in the anterior SNpc, where the most

susceptible neuronal population is located. Reactive astrocytes probably participate in

endogenous cell repair or neuroprotective mechanisms triggered at very early times following

exposure to the toxin, possibly involving release of brain-derived neurotrophic factor, glial

cell-line derived neurotrophic factor, and nerve growth factor, among many others.

From epidemiological data it is suggested that lifestyle might influence PD

etiology and progression. The risk of PD might be influenced, for example,

by educational achievement or occupation as well as by exercise (Frigerio

et al. 2005; Thacker et al. 2008). The use of enriched environments in

experimental situations can model both intellectual stimulation and

physical activity.

4.7 Astrocyte influence on neurogenesis

It is known that there are progenitor cells in the SN, from which new neurons can be formed

(Lie et al. 2002). Stimulating proliferation and differentiation would be very useful to replace

the neurons that are lost in PD. Finding the optimal conditions for this would also help in

potential stem cell replacement therapies. Astrocytes might help in this process by producing

various neurotrophic factors, synthesising extracellular substrates for axonal outgrowth and

synaptogenesis, and providing structural support and guiding migration. They can even act as

astrocytic progenitors during long-term recovery after brain injury (Liberto et al. 2004).

Astrocytes might need to become reactive to induce these properties. It has been found that

mesencephalic progenitor cells survive and differentiate better in rat PD models, than in

26

Page 27: Astrocytes in Parkinson's disease

normal rats (Sun et al. 2003), and extracts from dopamine-depleted striatum show a stronger

trophic activity (Nakajima et al. 2001). Reactive astrocytes might mediate increased basic

fibroblast growth factor and glial cell-line derived neurotrophic factor levels, or other

neurotrophic factors. This can create an environment in which proliferation and differentiation

of progenitor cells is stimulated which can potentially be modulated to create new therapies

(Chen et al. 2005).

4.8 Nrf2-mediated gene transcription inducing a large neuroprotective response

As discussed before, it is known that oxidative stress might be an important cause of neuronal

degradation in PD. An endogenous cellular defence mechanism against oxidative stress is the

binding of the transcription factor nuclear factor E2-related factor 2 (Nrf2) to the antioxidant

response element (ARE) enhancer sequence. This activates many antioxidant and anti-

inflammatory genes as well as growth factors, inducing a large neuroprotective response.

Nrf2-ARE activated genes include HO-1, NAD(P)H quinone oxidoreductase-1 and

glutathione S-transferases as well as glutathione-synthesizing enzymes

glutamate-cysteine ligase catalytic subunit and glutamate-cysteine ligase

modifier subunit (resulting in increased GSH levels). Inflammatory

mediators that are downregulated by Nrf2 include iNOS and COX-2. Nrf2 is

normally bound to its cytosolic suppressor Keap1, which dissociates in

response to oxidative stress, allowing Nrf2 to translocate to the nucleus

and induce transcription. Nrf2 is a very general defense mechanism but in

the brain it is believed that this response is mainly activated in astrocytes, since over

expression of Nrf2 in astrocytes is sufficient to prevent MPTP-induced neuronal cell death

(Chen et al. 2009). Induction of this pathway has also been found to reduce astrogliosis

(Kanninen et al. 2009). Moreover, Nrf2 translocates to the nucleus after microglial exposure

to astrocyte cultured medium, indicating that reactive astrocytes might be able to induce this

pathway in other cells (Min et al. 2006).

27

Page 28: Astrocytes in Parkinson's disease

5. Potential therapies for PD treatment targeting astrocytes

Some of these findings about the role of astrocytes in PD can be used to design new PD

therapies, targeting astrocytes. The neuroinflammatory hypothesis implies that drugs with an

anti-inflammatory mode of action should either arrest, or effectively slow down the

neurodegenerative disease progression. The pursuit of novel molecular and cellular targets

could be used for future anti-inflammatory drug development. Establishing which of the

inflammatory mechanisms are the most powerful in sustaining inflammation, and finding

methods to reduce their effects, might be the key to developing truly effective therapy.

It has been found that non-aspirin NSAID use is associated with lower risk of PD, though

there might be gender differences in this protective effect (Hernan et al. 2006). This clearly

indicates that inflammation is involved, and reducing inflammation can have a positive effect.

This effect could be due to classical NSAID action on cyclooxygenases (COXs) or their effect

on other COX independent targets such as transcription factors NF-κB, NO synthase, and

others (Asanuma & Miyazaki 2007).

Lee et al. (2009) found that H2S can reduce production of inflammatory mediators by

astrocytes and had neuroprotective effects through its antioxidant activity. This indicates there

is a considerable therapeutic potential for H2S releasing drugs in the treatment of

neurodegenerative disorders characterised by inflammatory processes, such as PD. Damaging

effects of inflammation could be reduced by supplementary H2S provided by drugs. H2S

releasing NSAIDS such as S-aspirin and S-diclofenac attenuate the neuroinflammation

induced by activation of astrocytes. However, consequent actions on blood vessels, where

H2S is also synthesised, must be taken into account.

Min et al. (2006) showed that astrocytes can regulate microglia activity by inducing HO-1.

They also found that mimickers of HO-1 products, such as bilirubin, ferrous iron, and a

carbon monoxide-releasing molecule, reduced interferon-γ-induced iNOS expression and/or

NO release in microglia. Such components could be used as potential therapies to control the

microglial inflammatory response. It has been found for example that overexpression of Nrf2,

which induces HO-1, can protect again 6-OHDA (Jakel et al. 2007) as well at MPTP induced

damage (Chen et al. 2009) possibly through HO-1 antioxidant and microglia regulatory

effects.

Another factor that can modulate astrocyte activity,, and could be a potential therapeutic

agent, is ONO-2506 (Kato et al. 2003). This substance inhibits the expression of COX-2 or

iNOS mRNA, induced in activated astrocytes. In the MPTP-mouse model, ONO-2506

28

Page 29: Astrocytes in Parkinson's disease

treatment prevented reduction in striatal dopamine and loss of loss of dopaminergic neurons

in the SN. Pre-treatment with this drug has no effect, indicating that is does not influence

MPTP toxicity but acts on astrocytes which are active later. ONO-2506 acts selectively on

astrocytes and modulates their activation or prevents too much activation that may be harmful

to neighbouring neurons. Interestingly, in ONO-2506 treated mice there is no reactive

astrogliosis peaking at 7 days after MPTP-treatment, as in normal situations, but there is a

moderate activation of astrocytes 3 days after MPTP treatment. This suggests that astrocytic

activation is facilitated, but to a limited degree, promoting only protective effects (Kato et al.

2003).

A very promising therapeutic target is the Nrf2-ARE pathway, which can induce many

different protective genes at once. Increased Nrf2-mediated gene transcription can be

achieved by overexpression of Nrf2 via injection of lentiviral or other vectors containing this

gene, into the SN (Kanninen et al. 2009). This can be done specifically in astrocytes using an

astrocyte promoter, or in all cells. Transgene expression then induces specific protection

against oxidative stress and reduces inflammation, which might help prevent neuronal

degradation in the SN. Another possibility is the oral administration of synthetic triterpenoids

which reduce Keap1 binding to Nrf2, increasing Nrf2 translocation to the nucleus, thus also

increasing expression of Nrf2-ARE regulated antioxidant and anti-inflammatory genes (Yang

et al. 2009).

There are many more factors involved in astrogliosis that could be manipulated to control the

astroglial reaction, an overview of these is given in Buffo et al. 2009. Of course, manipulation

of astrogliosis remains tricky, as astrocyte response is different in different neuropathologies

and certain effects might be beneficial as well as detrimental depending on the specific

timing.

29

Page 30: Astrocytes in Parkinson's disease

6. Discussion

Astrocytes play an important role in neurodegenerative diseases. In situations of neuronal

damage they can become reactive, a process which is called reactive astrogliosis (Eddleston &

Mucke 1993). This can be both beneficial, helping with neuronal repair or it can help sustain

inflammation, which could lead to more damage. Astrocytes react differently to different

types of damage and signals from neighbouring glial or neuronal cells. On top of this,

astrocytes form a heterogeneous population and can have different characteristics in different

brain areas, influencing their response to damage.

From post mortem studies and studies in animal models of PD, it is suggested that astrocytes

also play a role in PD (Kohutnica et al. 1998; Sheng et al. 1993). However, not much is

known about the specific mechanisms involved. It might be that astrocytes in the SN are

particularly vulnerable to certain types of damage or specific mutations, triggering neuronal

cell death in this area and inducing PD (Mena & Yebenes 2008; Morga et al. 1998). Or, once

astrogliosis is started, astrocytes can sustain and propagate inflammation (Liberatore et al.

1999; Miklossy et al. 2006). On the other hand, the reactive state of astrocytes might help

facilitate repair.

There are several factors that might have detrimental effects in PD. Mutated α-syn in

astrocytes can induce inflammation and aggregated forms of this protein cause neuronal cell

death (Klegeris et al. 2006; Stefanova et al. 2001; Wakabayashi et al. 2000). ER stress and

abberant protein folding are two more factors implicated in PD that can lead to cell death

(Slodzinksi et al. 2009). In their reactive state, astrocytes might also loose functions that are

important for normal neuronal survival such as the ability to regulate extracellular glutamate

levels (Dervan et al. 2004). Increased MAO-B levels in astrocytes due to age or disease result

in increased production of reactive oxygen species, contributing to neuronal cell loss (Damier

et al. 1996; Mallajosyula et al. 2008). ROS production can also be induced by inflammatory

mediators (Bolanos et al. 1994). Normally this is counteracted by increased GSH expression

and efflux but this might be a transient mechanism providing only limited protection (Chen et

al. 2001; Heales et al. 2004). Upregulation of ICAM-1 in the SN of PD patients indicates that

inflammation in this area might be self sustaining (Miklosssy et al. 2006). It is not known why

neurodegenerative effects are sometimes larger than protective effects but this could have

something to do with early exposure to hormones, programming astrocytes for a certain

response (Marchetti et al. 2005).

30

Page 31: Astrocytes in Parkinson's disease

Protective effects might be mediated through upregulation of PAR-1 which increases GSH

expression and secretion of neurotrophic factors (Ishida et al. 2006). Nurr 1 reduces

inflammation via communication with microglia (Saijo et al. 2009). Antioxidants such as H2S

can help clean up ROS produced during inflammation (Lee et al. 2009). Adenosine might

have protective effects through its stimulation of astrocyte proliferation (Ciccarelli et al. 2001;

Jenner et al. 2009). Enriched environment in animal models or possibly lifestyle in patients

can also protect against neuronal damage presumably via induction of an early protective

astrocytic response (Anastasia et al. 2009). Finally, astrocytes can stimulate neurogenesis via

various neurotrophic factors (Chen et al. 2005) and protect against oxidative stress via

induction of Nrf2-mediated transcription of antioxidant genes (Chen et al. 2009; Kanninen et

al. 2009).

Many of the factors increase reactive astrogliosis but this can have negative as well as positive

effects. Potential new PD therapies aimed at astrocytes have to find a way to distinguish

between these two effects. Inhibiting astrogliosis will suppress neurodegenerative but also

many protective effects and necessary homeostasis maintaining functions of astrocytes.

Protective effects should be stimulated without simultaneously stimulating degenerative

effects, or degenerative effects should be blocked without inhibiting protective effects. This

requires specific targeting of receptors and pathways involved in these processes. Some

examples are given of potential therapies aimed at astrocyte functioning. However, much

better understanding of the role of astrocytes in PD and the molecular mechanisms underlying

their effects is needed to find specific therapies that can inhibit the induction or progression of

this disease. Especially important would be to find what triggers or initiates the disease so the

disease can be recognised more early, and treated from the start, before too many neurons are

lost. A promising treatment is also the stimulation of neurogenesis, which might even be able

to repair neuronal networks to some extent. A possible way to achieve this would be to

manipulate astrocytes in such a way that they favour the survival and differentiation of stem

cell transplants and/or natural progenitor cells in the SN (Chen et al. 2005).

A very promising treatment is also the induction of Nrf2-mediated gene transcription (Chen et

al. 2009; Kanninen et al. 2009; Yang et al. 2009). This activates a large general

neuroprotective response through the regulation of antioxidant and anti-inflammatory genes

and growth factors. Instead of targeting single molecules and genes this is a relatively easy

way to influence many genes at the same time. Activation of Nrf2 gene transcription has

already been shown to protect against neuronal cell loss in the MPTP model (Yang et al.

2009). Increasing the neuroprotective and antioxidant potential of astrocytes, via induction of

31

Page 32: Astrocytes in Parkinson's disease

the Nrf2-ARE pathway seems to be a very effective way to prevent neuronal degradation in

the SN and halt PD progression.

Astrocytes are a very promising target for the treatment of PD. Conventional therapies which

are mostly aimed at neurons, are still not able to stop the progression of the disease or reduce

the symptoms for a longer period of time. A problem with therapies targeting neurons is that

as long as neuronal death is not prevented, eventually there are not enough neurons left to

manipulate. Astrocytes on the other hand only increase their numbers when neuronal cell

death increases, and remain in the area. Astrocytes can be manipulated to stop or slow down

disease progression and possibly even stimulate repair at later stages of the disease, when

many neurons have already died, and other treatments loose effectivity. Therefore, it is very

important to continue research into astrocyte function in PD pathology, to get a better

understanding of the mechanisms underlying the disease and find possible therapeutic targets.

32

Page 33: Astrocytes in Parkinson's disease

List of abbreviations

6-OHDA 6-hydroxydopamine

α-syn α-synuclein

ACM astrocytic culture medium

ARE antioxidant response element

CBS cystathionine-b-synthase

CNS central nervous system

COX cyclooxygenase

DA dopamine

DAT dopamine active transporter

EE enriched environment

GABA γ-aminobyturic acid

GCL glutamate-cysteine ligase

GP globus pallidus

GSH glutathione

HO-1 heme oxygenase 1

IFN-γ interferon γ

IL-6 interleukin-6

ICAM-1 intracellular adhesion molecule 1

iNOS inducible nitric oxide synthetase

l-dopa levodopa

LFA-1 lymphocyte function-associated antigen 1

LPS lipopolysacharide

MAO-B monoamine oxidase B

MAPK mitogen activated protein kinase

MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

MPP+ 1-methyl-4-phenylpyridinium

NF-κB nuclear factor κ B

Nrf2 NF-E2-related factor 2

NO nitric oxide

NSAID non steroidal anti inflammatory drug

NQO1 NADPH quinone oxidoreductase 1PAR-1 protease activated receptor 1

PD Parkinson’s disease

SN(pc) Substantia nigra (pars compacta)

STN subthalamic nucleus

TH tyrosine hydroxylase

TNFα tumor necrosis factor α

WT wild type

33

Page 34: Astrocytes in Parkinson's disease

References

Aderka D, Le JM, Vilcek J. IL-6 inhibits lipopolysaccharide-induced tumor necrosis factor production in cultured human monocytes, U937 cells, and in mice. J Immunol. 1989 Dec 1;143(11):3517-23.

Anastasía A, Torre L, de Erausquin GA, Mascó DH. Enriched environment protects the nigrostriatal dopaminergic system and induces astroglial reaction in the 6-OHDA rat model of Parkinson's disease. J Neurochem. 2009 May;109(3):755-65.

Aponso PM, Faull RL, Connor B. Increased progenitor cell proliferation and astrogenesis in the partial progressive 6-hydroxydopamine model of Parkinson's disease. Neuroscience. 2008 Feb 19;151(4):1142-53

Asanuma M, Miyazaki I. Common anti-inflammatory drugs are potentially therapeutic for Parkinson's disease? Exp Neurol. 2007 Aug;206(2):172-8.

Bachoo RM, Kim RS, Ligon KL, Maher EA, Brennan C, Billings N, Chan S, Li C, Rowitch DH, Wong WH, DePinho RA. Molecular diversity of astrocytes with implications for neurological disorders. Proc Natl Acad Sci U S A. 2004 Jun 1;101(22):8384-9.

Bolaños JP, Peuchen S, Heales SJ, Land JM, Clark JB. Nitric oxide-mediated inhibition of the mitochondrial respiratory chain in cultured astrocytes. J Neurochem. 1994 Sep;63(3):910-6.

Bove J, Prou D, Perier C, Przedborski S. Toxin-induced models of Parkinson’s disease. NeuroRx 2005; 2:484–494

Brahmachari S, Fung YK, Pahan K. Induction of glial fibrillary acidic protein expression in astrocytes by nitric oxide. J Neurosci. 2006 May 3;26(18):4930-9.

Buffo A, Rolando C, Ceruti S. Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential. Biochem Pharmacol. 2010 Jan 15;79(2):77-89. Epub 2009 Sep 16.

Bushong EA, Martone ME, Jones YZ, Ellisman MH. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains.J. Neurosci. 2002; 22, 183–192.

Chen LW, Yung KL, Chan YS. Reactive astrocytes as potential manipulation targets in novel cell replacement therapy of Parkinson's disease. Curr Drug Targets. 2005 Nov;6(7):821-33.

Chen PC, Vargas MR, Pani AK, Smeyne RJ, Johnson DA, Kan YW, Johnson JA. Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson's disease: Critical role for the astrocyte. Proc Natl Acad Sci U S A. 2009 Feb 24;106(8):2933-8.

Chen Y, Vartiainen NE, Ying W, Chan PH, Koistinaho J, Swanson RA. Astrocytes protect neurons from nitric oxide toxicity by a glutathione-dependent mechanism. J Neurochem. 2001 Jun;77(6):1601-10

Chesselet MF, Fleming S, Mortazavi F, Meurers B. Strengths and limitations of genetic mouse models of Parkinson's disease. Parkinsonism Relat Disord. 2008;14 Suppl 2:S84-7.

Ciccarelli R, Di Iorio P, Giuliani P, D’Alimonte I, Ballerini P, Caciagli F, Rathbone MP. Rat cultured astrocytes release guanine-based purines in basal conditions and after hypoxia/hypoglycemia. Glia 1999; 25, 93–98.Ciccarelli R, Ballerini P, Sabatino G, Rathbone MP, D'Onofrio M, Caciagli F, Di Iorio P. Involvement of

astrocytes in purine-mediated reparative processes in the brain. Int J Dev Neurosci. 2001 Jul;19(4):395-414.Cohen G, Farooqui R, Kesler N. Parkinson disease: a new link between monoamine oxidase and mitochondrial

electron flow. Proc Natl Acad Sci U S A. 1997 May 13;94(10):4890-4.Croisier E, Graeber MB. Glial degeneration and reactive gliosis in alpha-synucleinopathies: the emerging

concept of primary gliodegeneration. Acta Neuropathol. 2006 Nov;112(5):517-30.Damier P, Hirsch EC, Zhang P, Agid Y, Javoy-Agid F. Glutathione peroxidase, glial cells and Parkinson's

disease. Neuroscience. 1993 Jan;52(1):1-6.Damier P, Kastner A, Agid Y, Hirsch EC. Does monoamine oxidase type B play a role in dopaminergic nerve

cell death in Parkinson's disease? Neurology. 1996 May;46(5):1262-9.de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M, Fratiglioni L, Lobo A, Martinez-

Lage J,Trenkwalder C, Hofman A. Prevalence of Parkinson's disease in Europe: A collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology.  2000;54(11 Suppl 5):S21-3

Depino AM, Earl C, Kaczmarczyk E, Ferrari C, Besedovsky H, del Rey A, Pitossi FJ, Oertel WH. Microglial activation with atypical proinflammatory cytokine expression in a rat model of Parkinson's disease. Eur J Neurosci. 2003 Nov;18(10):2731-42

Dervan AG, Meshul CK, Beales M, McBean GJ, Moore C, Totterdell S, Snyder AK, Meredith GE. Astroglial plasticity and glutamate function in a chronic mouse model of Parkinson's disease. Exp Neurol. 2004 Nov;190(1):145-56.

Eddleston M, Mucke L. Molecular profile of reactive astrocytes--implications for their role in neurologic disease. Neuroscience. 1993 May;54(1):15-36.

34

Page 35: Astrocytes in Parkinson's disease

Eng LF, Ghirnikar RS, Lee YL. Glial fibrillary acidic protein: GFAP-thirty-one years (1969-2000). Neurochem Res. 2000 Oct;25(9-10):1439-51.

Fattori E, Lazzaro D, Musiani P, Modesti A, Alonzi T, Ciliberto G. IL-6 expression in neurons of transgenic mice causes reactive astrocytosis and increase in ramified microglial cells but no neuronal damage. Eur J Neurosci. 1995 Dec 1;7(12):2441-9.

Follmer C, Romão L, Einsiedler CM, Porto TC, Lara FA, Moncores M, Weissmüller G, Lashuel HA, Lansbury P, Neto VM, Silva JL, Foguel D. Dopamine affects the stability, hydration, and packing of protofibrils and fibrils of the wild type and variants of alpha-synuclein. Biochemistry. 2007 Jan 16;46(2):472-82.

Frei K, Malipiero UV, Leist TP, Zinkernagel RM, Schwab ME, Fontana A. On the cellular source and function of interleukin 6 produced in the central nervous system in viral diseases. Eur J Immunol. 1989 Apr;19(4):689-94.

Frigerio R, Elbaz A, Sanft K, Peterson B, Bower J, Ahlskog J, Grossardt B, De Andrade M, Maraganore D, Rocca W. Education and occupations preceding Parkinson disease: apopulation-based case–control study. Neurology 2005; 65, 1575–1583.

Gasser T. Molecular pathogenesis of Parkinson disease: insights from genetic studies. Expert Rev Mol Med. 2009 Jul 27;11:e22.

Gegg ME, Beltran B, Salas-Pino S, Bolanos JP, Clark JB, Moncada S, Heales SJ. Differential effect of nitric oxide on glutathione metabolism and mitochondrial function in astrocytes and neurones: implications for neuroprotection/neurodegeneration? J Neurochem. 2003 Jul;86(1):228-37.

Gelb DJ, Oliver E, Gilman S. Diagnostic criteria for Parkinson disease. Arch Neurol. 1999 Jan;56(1):33-9.Giulian D, Lachman LB. Interleukin-1 stimulation of astroglial proliferation after brain injury. Science. 1985

Apr 26;228(4698):497-9Halliday GM, Del Tredici K, Braak H. Critical appraisal of brain pathology staging related to presymptomatic

and symptomatic cases of sporadic Parkinson's disease. J Neural Transm Suppl. 2006;(70):99-103. Harvey BK, Wang Y, Hoffer BJ. Transgenic rodent models of Parkinson's disease. Acta Neurochir Suppl.

2008;101:89-92Heales SJ, Lam AA, Duncan AJ, Land JM. Neurodegeneration or neuroprotection: the pivotal role of astrocytes.

Neurochem Res. 2004 Mar;29(3):513-9.Henning J, Strauss U, Wree A, Gimsa J, Rolfs A, Benecke R, Gimsa U. Differential astroglial activation in 6-

hydroxydopamine models of Parkinson's disease. Neurosci Res. 2008 Dec;62(4):246-53. Hernán MA, Logroscino G, García Rodríguez LA. Nonsteroidal anti-inflammatory drugs and the incidence of

Parkinson disease. Neurology. 2006 Apr 11;66(7):1097-9.Hirsch EC, Breidert T, Rousselet E, Hunot S, Hartmann A, Michel PP. The role of glial reaction and

inflammation in Parkinson's disease. Ann N Y Acad Sci. 2003 Jun;991:214-28Hunot S, Boissière F, Faucheux B, Brugg B, Mouatt-Prigent A, Agid Y, Hirsch EC. Nitric oxide synthase and

neuronal vulnerability in Parkinson's disease. Neuroscience. 1996 May;72(2):355-63.Ishida Y, Nagai A, Kobayashi S, Kim SU. Upregulation of protease-activated receptor-1 in astrocytes in

Parkinson disease: astrocyte-mediated neuroprotection through increased levels of glutathione peroxidase. J Neuropathol Exp Neurol. 2006 Jan;65(1):66-77.

Jakel RJ, Townsend JA, Kraft AD, Johnson JA. Nrf2-mediated protection against 6-hydroxydopamine. Brain Res. 2007 May 4;1144:192-201.

Jenner P, Mori A, Hauser R, Morelli M, Fredholm BB, Chen JF. Adenosine, adenosine A 2A antagonists, and Parkinson's disease. Parkinsonism Relat Disord. 2009 Jul;15(6):406-13.

Kanninen K, Heikkinen R, Malm T, Rolova T, Kuhmonen S, Leinonen H, Ylä-Herttuala S, Tanila H, Levonen AL, Koistinaho M, Koistinaho J. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A. 2009 Sep 22;106(38):16505-10.

Kato H, Araki T, Imai Y, Takahashi A, Itoyama Y. Protection of dopaminergic neurons with a novel astrocyte modulating agent (R)-(-)-2-propyloctanoic acid (ONO-2506) in an MPTP-mouse model of Parkinson's disease. J Neurol Sci. 2003 Apr 15;208(1-2):9-15.

Kirik D, Bjorklund A. Modeling CNS neurodegeneration by overexpression of disease-causing proteins using viral vectors. Trends Neurosci 2003;26:386–392.

Klegeris A, Giasson BI, Zhang H, Maguire J, Pelech S, McGeer PL. Alpha-synuclein and its disease-causing mutants induce ICAM-1 and IL-6 in human astrocytes and astrocytoma cells. FASEB J. 2006 Oct;20(12):2000-8.

Klegeris A, McGeer EG, McGeer PL. Therapeutic approaches to inflammation in neurodegenerative disease. Curr Opin Neurol. 2007 Jun;20(3):351-7.

Klegeris A, Pelech S, Giasson BI, Maguire J, Zhang H, McGeer EG, McGeer PL. Alpha-synuclein activates stress signaling protein kinases in THP-1 cells and microglia. Neurobiol Aging. 2008 May;29(5):739-52.

35

Page 36: Astrocytes in Parkinson's disease

Kohutnicka M, Lewandowska E, Kurkowska-Jastrzebska I, Członkowski A, Członkowska A. Microglial and astrocytic involvement in a murine model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). mmunopharmacology. 1998 Jun;39(3):167-80.

Kumar MJ, Nicholls DG, Andersen JK. Oxidative alpha-ketoglutarate dehydrogenase inhibition via subtle elevations in monoamine oxidase B levels results in loss of spare respiratory capacity: implications for Parkinson's disease. J Biol Chem. 2003 Nov 21;278(47):46432-9

Kumar MJ, Andersen JK. Perspectives on MAO-B in aging and neurodegenerative disease: where do we go from here? Mol Neurobiol. 2004 Aug; 30(1):77-89

Le WD, Xu P, Jankovic J, Jiang H, Appel SH, Smith RG, Vassilatis DK. Mutations in NR4A2 associated with familial Parkinson disease. Nat. Genet. 2003; 33, 85–89.

Lee HJ, Patel S, Lee SJ. Intravesicular localization and exocytosis of alpha-synuclein and its aggregates. J Neurosci. 2005 Jun 22;25(25):6016-24.

Lee M, Schwab C, Yu S, McGeer E, McGeer PL. Astrocytes produce the antiinflammatory and neuroprotective agent hydrogen sulfide. Neurobiol Aging. 2009 Oct;30(10):1523-34.

Lee VM, Trojanowski JQ. Mechanisms of Parkinson's disease linked to pathological alpha-synuclein: new targets for drug discovery. Neuron. 2006 Oct 5;52(1):33-8.

Liberatore GT, Jackson-Lewis V, Vukosavic S, Mandir AS, Vila M, McAuliffe WG, Dawson VL, Dawson TM, Przedborski S. Inducible nitric oxide synthase stimulates dopaminergic neurodegeneration in the MPTP model of Parkinson disease. Nat Med. 1999 Dec;5(12):1403-9.

Liberto CM, Albrecht PJ, Herx LM, Yong VW, Levison SW. Pro-regenerative properties of cytokine-activated astrocytes. J. Neurochem. 2004; 89, 1092-1100.Lie DC, Dziewczapolski G, Willhoite AR, Kaspar BK, Shults CW, Gage FH. The adult substantia nigra contains

progenitor cells with neurogenic potential. J Neurosci. 2002 Aug 1;22(15):6639-49.Liu B. Modulation of microglial pro-inflammatory and neurotoxic activity for the treatment of Parkinson's

disease. AAPS J. 2006 Sep 29;8(3):E606-21.Mallajosyula JK, Kaur D, Chinta SJ, Rajagopalan S, Rane A, Nicholls DG, Di Monte DA, Macarthur H,

Andersen JK. MAO-B elevation in mouse brain astrocytes results in Parkinson's pathology. PLoS One. 2008 Feb 20;3(2):e1616.

Marchetti B, Serra PA, L'Episcopo F, Tirolo C, Caniglia S, Testa N, Cioni S, Gennuso F, Rocchitta G, Desole MS, Mazzarino MC, Miele E, Morale MC. Hormones are key actors in gene x environment interactions programming the vulnerability to Parkinson's disease: glia as a common final pathway. Ann N Y Acad Sci. 2005 Dec;1057:296-318.

McGeer PL, McGeer EG. Glial reactions in Parkinson's disease. Mov Disord. 2008 Mar 15;23(4):474-83.Melrose HL, Lincoln SJ, Tyndall GM, Farrer MJ. Parkinson's disease: a rethink of rodent models. Exp Brain

Res. 2006 Aug;173(2):196-204. Epub 2006 Apr 26.Mena MA, García de Yébenes J. Glial cells as players in parkinsonism: the "good," the "bad," and the

"mysterious" glia. Neuroscientist. 2008 Dec;14(6):544-60.Meredith GE, Totterdell S, Petroske E, Santa Cruz K, Callison RC Jr, Lau YS. Lysosomal malfunction

accompanies alpha-synuclein aggregation in a progressive mouse model of Parkinson's disease. Brain Res. 2002 Nov 22;956(1):156-65.

Miklossy J, Doudet DD, Schwab C, Yu S, McGeer EG, McGeer PL. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Exp Neurol. 2006 Feb;197(2):275-83.

Min KJ, Jou I, Joe E. Plasminogen-induced IL-1beta and TNF-alpha production in microglia is regulated by reactive oxygen species. Biochem Biophys Res Commun 2003; 312:969 –974.Min KJ, Yang MS, Kim SU, Jou I, Joe EH. Astrocytes induce hemeoxygenase-1 expression in microglia: a

feasible mechanism for preventing excessive brain inflammation. J Neurosci. 2006 Feb 8;26(6):1880-7.Mirza B, Hadberg H, Thomsen P, Moos T. The absence of reactive astrocytosis is indicative of a unique

inflammatory process in Parkinson's disease. Neuroscience. 2000;95(2):425-32.Moratalla R, Quinn B, DeLanney LE, Irwin I, Langston JW, Graybiel AM (1992) Differential vulnerability of

primatecaudate-putamen and striosome-matrix dopamine systems to the neurotoxic effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad Sci USA 89:3859–3863

Morga E, Faber C, Heuschling P. Cultured astrocytes express regional heterogeneity of the immunoreactive phenotype under basal conditions and after gamma-IFN induction. J Neuroimmunol. 1998 Jul 1;87(1-2):179-84.

Muthane U, Ramsay KA, Jiang H, Jackson-Lewis V, Donaldson D, Fernando S, Ferreira M, Przedborski S (1994) Differences in nigral neuron number and sensitivity to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in C57/bl and CD-1mice. Exp Neurol 126:195–204

Nakajima K, Hida H, Shimano Y, Fujimoto I, Hashitani T,Kumazaki M, Sakurai T, Nishino H. GDNF is a major component of trophic activity in DA-depleted striatum for survival and neurite extension of DAergic neurons. Brain Res. 2001; 916:76-84.

36

Page 37: Astrocytes in Parkinson's disease

Norris EH, Giasson BI, Lee VM. Alpha-synuclein: normal function and role in neurodegenerative diseases. Curr Top Dev Biol. 2004;60:17-54.

Norton WT, Aquino DA, Hozumi I, Chiu FC, Brosnan CF. Quantitative aspects of reactive gliosis: a review. Neurochem Res. 1992 Sep;17(9):877-85.

Orr CF, Rowe DB, Halliday GM. An inflammatory review of Parkinson's disease. Prog Neurobiol. 2002 Dec;68(5):325-40.

Pearce RK, Owen A, Daniel S, Jenner P, Marsden CD. Alterations in the distribution of glutathione in the substantia nigra in Parkinson's disease. J. Neural. Transm. 1997 104(6-7):661-77

Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005 Jun;50(4):427-34.Purves D, Augustine GJ, Fitzpatrick D, Katz LC, LaMantia AS, McNamara JO, Williams SM. Neuroscience.

Sunderland (MA): Sinauer Associates, Inc. 2001 Ross OA, Farrer MJ. Pathophysiology, pleiotrophy and paradigm shifts: genetic lessons from Parkinson’s

disease. Biochem Soc Trans 2005; 33:586–590Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, Gage FH, Glass CK. A Nurr1/CoREST

pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell. 2009 Apr 3;137(1):47-59.

Seifert G, Schilling K, Steinhäuser C. Astrocyte dysfunction in neurological disorders: a molecular perspective. Nat Rev Neurosci. 2006 Mar;7(3):194-206.

Schapira AH, Emre M, Jenner P, Poewe W. Levodopa in the treatment of Parkinson's disease. Eur J Neurol. 2009 Sep;16(9):982-9

Sheng JG, Shirabe S, Nishiyama N, Schwartz JP. Alterations in striatal glial fibrillary acidic protein expression in response to 6-hydroxydopamine-induced denervation. Exp Brain Res. 1993;95(3):450-6

Sian J, Dexter DT, Lees AJ, Daniel S, Agid Y, Javoy-Agid F, Jenner P, Marsden CD. Alterations in glutathione levels in Parkinson's disease and other neurodegenerative disorders affecting basal ganglia. Ann Neurol. 1994 Sep;36(3):348-55.

Slodzinski H, Moran LB, Michael GJ, Wang B, Novoselov S, Cheetham ME, Pearce RK, Graeber MB. Homocysteine-induced endoplasmic reticulum protein (herp) is up-regulated in parkinsonian substantia nigra and present in the core of Lewy bodies. Clin Neuropathol. 2009 Sep-Oct;28(5):333-43.

Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009 Sep 24. [Epub ahead of print]

Sriram K, Benkovic SA, Hebert MA, Miller DB, O'Callaghan JP. Induction of gp130-related cytokines and activation of JAK2/STAT3 pathway in astrocytes precedes up-regulation of glial fibrillary acidic protein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of neurodegeneration: key signaling pathway for astrogliosis in vivo? J Biol Chem. 2004 May 7;279(19):19936-47.

Stefanova N, Klimaschewski L, Poewe W, Wenning GK, Reindl M. Glial cell death induced by overexpression of alpha-synuclein. J Neurosci Res. 2001 Sep 1;65(5):432-8

Sun ZH, Lai YL, Zeng WW, Zhao D, Zuo HC, Xie ZP. Mesencephalic progentitors can improve rotational behaviour and reconstruct nigrostriatal pathway in PD rats. Acta Neurochir. Suppl., 2003 ; 87, 169-174.Teismann P, Schulz JB. Cellular pathology of Parkinson's disease: astrocytes, microglia and inflammation. Cell

Tissue Res. 2004 Oct;318(1):149-61.Thacker EL, Chen H, Patel AV, McCullough ML, Calle EE, Thun MJ, Schwarzschild MA, Ascherio A.

Recreational physical activity and risk of Parkinson’s disease. Mov.Disord.2008; 23, 69–74.Vila M, Przedborski S. Targeting programmed cell death in neurodegenerative diseases. Nat Rev Neurosci. 2003

May;4(5):365-75.Volterra A, Meldolesi J. Astrocytes, from brain glue to communication elements: the revolution continues. Nat

Rev Neurosci. 2005 Aug;6(8):626-40. Wakabayashi K, Hayashi S, Yoshimoto M, Kudo H, Takahashi H. NACP/alpha-synuclein-positive filamentous

inclusions in astrocytes and oligodendrocytes of Parkinson's disease brains. Acta Neuropathol. 2000 Jan;99(1):14-20

Wang Z,Benoit G, Liu J, Prasad S, Aarnisalo P, Liu X, Xu H, Walker NP, Perlmann T. Structure and function of Nurr1 identifies a class of ligand-independent nuclear receptors. Nature 2003; 423, 555–560.

Watanabe Y, Himeda T, Araki T. Mechanisms of MPTP toxicity and their implications for therapy of Parkinson’s disease. Med Sci Monit 2005; 11:RA17–RA

Weidong Le, Shen Chen, Jankovic J. Etiopathogenesis of Parkinson disease: a new beginning? Neuroscientist. 2009 Feb;15(1):28-35.

Yang L, Calingasan NY, Thomas B, Chaturvedi RK, Kiaei M, Wille EJ, Liby KT, Williams C, Royce D, Risingsong R, Musiek ES, Morrow JD, Sporn M, Beal MF. Neuroprotective effects of the triterpenoid, CDDO methyl amide, a potent inducer of Nrf2-mediated transcription. PLoS One. 2009 Jun 1;4(6):e5757.

37